Abstract

Regenerative MedicineVol. 1, No. 4 EditorialFree AccessLarge animal models are critical for rationally advancing regenerative therapiesDustin R Wakeman, Andrew M Crain & Evan Y SnyderDustin R WakemanUniversity of California San Diego, Biomedical Sciences Graduate Program, 9500 Gilman Drive, La Jolla, CA 92093, USABurnham Institute for Medical Research, 10901 North Torrey Pines Rd, La Jolla, CA 92037, USA. Search for more papers by this authorEmail the corresponding author at esnyder@burnham.org, Andrew M CrainUniversity of California San Diego, Biomedical Sciences Graduate Program, 9500 Gilman Drive, La Jolla, CA 92093, USABurnham Institute for Medical Research, 10901 North Torrey Pines Rd, La Jolla, CA 92037, USA. Search for more papers by this authorEmail the corresponding author at esnyder@burnham.org & Evan Y Snyder† Author for correspondenceBurnham Institute for Medical Research, 10901 North Torrey Pines Rd, La Jolla, CA 92037, USA. Search for more papers by this authorEmail the corresponding author at esnyder@burnham.orgPublished Online:18 Jul 2006https://doi.org/10.2217/17460751.1.4.405AboutSectionsPDF/EPUB ToolsAdd to favoritesDownload CitationsTrack CitationsPermissionsReprints ShareShare onFacebookTwitterLinkedInReddit Keywords: GDNFgene therapylarge animal modelMPTPnonhuman primateParkinson’s diseasestem/precursor celltransplantationType 1 diabetesregenerative medicineEnthusiasm for therapies based on the transplantation of exogenous cells or the transfer of genes by viral vectors has burgeoned over the past 30 years, accompanied by a predictable exhortation to launch clinical trials as soon as possible. Most data regarding safety, efficacy and mechanisms of these therapies have been derived from studies in rodents alone. While such ‘small-animal’ systems offer invaluable insights into fundamental biological questions, it is often misleading and perilous to unquestionably equate the higher order motor, sensory and cognitive processes that characterize human disease with that gleaned from a mouse or rat. Indeed, the literature is littered with clinical trials that failed and, in some cases, led to unforeseen adverse outcomes because the field leap-frogged over the requisite large-animal model. Large animals often provide an essential bridge between insights into fundamental biology and pathophysiology gleaned from simple systems and the realities of treating a human disease. Often, this is especially true for neurological disorders where not only differences in size and scale pertain, but also in neuroanatomical connections and organization, cognitive capacities, signaling pathways, genetic redundancy or the disease etiology.While the gene therapy field has increased their use of nonhuman primates prior to the application of viral vectors in clinical trials, the cellular therapy field – represented most conspicuously of late by the stem cell field – has only recently begun to properly address this requirement. Monkeys and the minipig may prove to be excellent preclinical models owing to their similar comparative anatomy, pharmacokinetics and physiological and metabolic interactions. These models have proven to be extremely useful for studying endocrinological diseases, such as Type 1 diabetes, and neurological disorders, including Parkinson’s disease (PD), spinal cord injury (SCI) and multiple sclerosis (MS). Prudence would argue that clinical trials for diseases in which differences between rodent and human transcend size and scale should require clear and definitive proof-of-concept in at least one relevant large-animal model in order to safeguard patients. This need, of course, is counterbalanced by considerations of the substantial cost, time and ethical circumspection that typically accompanies such research. In this editorial, we will attempt to help researchers reason through the potential need and advisability of using a large-animal model for their particular biological question. There are numerous differences between rodents and humans that make the unqualified translation of rodent data inadequate. The most relevant differences start at the genomic level and are manifested as differences in pharmacological, biochemical, developmental, behavioral and functional responses to perturbations and interventions [1]. Anatomical and cytoarchitectural differences are particularly profound in the brain, where innervation of specific regions is critical to addressing a neurological deficit (Figure 1). In PD for example, it has been estimated that for each volume of tissue innervated by a rat dopaminergic (DA) neuron, a monkey neuron must innervate 20-times that volume, while humans would need a staggering volume of 200 units to mimic similar reinnervation of nigrastriatal projections [2]. In addition, differences in monoamine biochemistry and developmental life span make extrapolating rodent data to human therapies risky and insufficient [3–7]. Behavioral differences between rodents and humans are also of great significance [8–10]. It is likely that only a large animal model can adequately mirror the complexities of studying and restoring bipedal gait, balance, tremor, hand preference, fine motor coordination, spontaneous blink rate and cognitive deficits [11–16]. Figure 1. Coronal images of Nissl stained sections from Mus musculus, Chlorocebus aethiops, Macaca mulatta, and MRI of human cortex [135].The minipigFor research into PD, the nonhuman primate model is ideal, although, the technical, financial and organizational complexities of monkey colony maintenance may require the use of an alternative, somewhat more accessible, species, such as the Gottingen miniature pig [17]. The ‘minipig’, developed in the early 1960s [18,19], presents many advantages over the rodent as a model for human diseases. Its organ systems are organized similar to that of the human and it displays similar physiological and pathophysiological responses [20]. Similarities in pancreas size, position and shape [21], gastrointestinal tract structure and function [20], and pharmacokinetic responses make the minipig an especially useful model for Type 1 diabetes [22]. The kinetics and dynamics after subcutaneous delivery of insulin, [23,24] as well as aspects of metabolism [25] and glucose tolerance [26,27], closely resemble those of humans. Furthermore, several stable models for chemical induction of Type 1 diabetes have been established in minipigs using pharmacological insult to ??cells with streptozotocin (STZ) [28–31] or alloxan [32]. Recently, the transplantation of both human and porcine islets of Langerhans in Type 1 diabetes has been studied in diabetic pigs, as well as in dogs, monkeys and patients [33–41].Complications in xenograft survival related to differences in the HLA and SLA systems between humans and pigs, respectively, are of concern, but may be partially overcome by using a more recent variation of the minipig – the ‘NIH minipig’. First described by Sachs in 1976 for transplantation studies [42,43], the NIH minipig, homozygous for known alleles of the major histocompatibility complex (MHC) [44], has been shown to be effective for modeling immunological and genetic aspects of heart, lung and spleen transplant tolerance [45,46,22] and is sufficiently bred to accept bone marrow and possibly hematopoetic stem cell transplants [47,48], as well as skin and heart grafts [22]. The minipig has a relatively large brain, approximately ten-times the mass of the marmoset [49,50], making it of recent interest in neurological disease studies. The size of the brain makes it especially appealing for imaging and stereotactic manipulation. There is a growing list of neurological diseases that have been modeled in the minipig: experimental allergic encephalomyelitis (EAE) [51], mimicking the autoimmune, demyelinating disease MS; Huntington’s disease (HD) CAG triplet repeats [52]; and 1?methyl?4?phenyl?1,2,3,6?tetrahydropyridine (MPTP)-induced Parkinsonism [53]. Such models enable a more representative analysis of cell transplantation therapies for these disorders. In fact, porcine xenografts have already been tested in Phase I trials in both HD and PD patients [54,55], as well as in a variety of other disorders [56,57], validating the use of the minipig as a bridge between rodent and human clinical trials. The minipig has also been used to study implants of progenitor cells into the ischemic and contused spinal cord (M Marsala; correspondence). Indeed, it is work in the spinal cord and the difficulties in attempting to adapt procedures developed in rodent to the pig that highlight the absolute need for large animal studies as a precursor to human trials for such disorders [58]. Lessons from Parkinson’s disease PD is characterized by degeneration of DA neurons in the substantia nigra pars compacta and subsequent loss of striatal DA release. Cell-based strategies for replacement of damaged DA neurons have been a promising strategy since the early 1970s, when Olsen and colleagues performed their initial transplantation experiments in rat brains [59–63]. Several subsequent studies in rats provided extended evidence for fetal cell transplants repairing DA deficiency [64,65]; however, despite numerous attempts by many groups, these results could never be repeated in primates [1]. Coupled with mounting ethical considerations, researchers turned to adrenal allografts as a new source of tissue; however, these also failed and subsequent clinical trials were abandoned [66–68]. New hope came again after 1985 when human fetal neural cells were derived and successfully implanted into the rat and monkey brain [69–76]; however ethical opponents argued that the large amount of fetal tissue required made these procedures for routine regenerative therapy unfeasible and, therefore, unfundable. Based on the varying methods published in the mid-1970s, several groups began preliminary efficacy and safety clinical studies of fetal ventral mesencephalic grafts in PD patients. Initial studies in Sweden [77], England [78] and Mexico [79] produced variable results, elevating the ethical debate and political controversy, eventually leading to a funding moratorium in the USA [80,81]. Subsequent studies appeared promising, although the overall consensus was that the results were inconclusive owing to variability within small studies, limited short-term functional improvements and overall difficulty in comparing independent procedures [1]. To investigate this problem and improve the ability to compare studies, a panel of experts published a recommended protocol for future investigations, termed Core Assessment Program for Intracerebral Transplantation (CAPIT) [82]. In the same year, the first randomized controlled clinical trial, headed by Curt Freed and Stanley Fahn, was initiated studying 40 PD patients over 7 years, half of whom underwent bilateral stereotactic transplantation of human embryonic mesencephalic tissue into the putamen [83]. Although initial results appeared promising, it was determined that no benefit in primary outcome measure had been attained after 12 months post operation. However, a small subset of patients under 60 years old did show small double-blind improvements on a validated PD rating scale. More importantly, predictions made from animal models failed to predict the relatively high occurrence of dystonia and dyskinesia associated with long-term survival [84]. These gross abnormalities occurred in five out of 33 patients that lived 3 years post transplant, of whom, all five were aged less than 60 years and had displayed improvements in PD score after 12 months post surgery. The human trials appeared to have a solid rationale based on a decade of rodent studies. Hundreds of investigators had reported survival and functional recovery from fetal ventral mesencephalic-derived neural tissue transplanted into the striatum of rats [85]. The grafts were shown to express tyrosine hydroxylase (TH), increase DA concentrations, project long outgrowths, form synaptic connections and respond to afferent stimulation [86–90]. However, the rat models of PD overestimated the likely success of fetal precursor transplants and failed to anticipate the dyskinetic side effects in human trials [84]. Furthermore, although large animal models were tested, it is possible that they were not followed for a long enough time, simply owing to the vast difference between rodents and primates (of which human is an example) was not appreciated. Also, while the Freed and colleagues study made extremely valuable contributions to our understanding of PD, small design flaws, such as method of tissue preparation, limited target area and lack of immunosuppression, may have inherently impeded the overall success of the study [1]. Many of these details may have been worked out in large animals before reaching the clinic, potentially predicting adverse effects. Unfortunately, cell replacement therapy received negative media coverage [91–94], creating an impression of defeat in the public’s mind and even creating dissention among PD investigators [84,92,95–102]. Much of this may have been avoided had a higher priority been placed on more extensive primate studies with larger numbers and longer follow-up.Going forward in Parkinson’s disease: a case-in-point for choosing the correct model Having detailed the pitfalls into which the PD field slipped by relying upon nonrepresentative data for its first cell-based clinical trial; how might it recover its momentum, and can this serve as a lesson for future trials for other diseases?As aforementioned, most of the early transplantation studies in PD were performed in rodents that had received a unilateral injection of the dopaminergic toxin 6?hydroxydopamine (6?OHDA) into the medial forebrain bundle or nearby [103,104]. Although transplantation appeared to promote recovery from the resultant PD-like symptoms, these behaviors actually bore little resemblance to human PD. If therapies were to be tested solely on such a rodent model, the results would likely have poor predictive value for actual patients. Conversely, through serendipity in the 1960s and 70s, it was discovered that the complex I inhibitor, MPTP, created pathology quite similar to human idiopathic PD in humans [105–107] and monkeys [70,108–112], especially the vervet [113,114]. MPTP depletes substantia nigra TH?immunoreactive and striatal DA to levels similar to PD, and produces characteristic Parkinsonian symptomatology, including akinesia, rigidity, postural alterations and tremor [9,10]. Overexpressing ??synuclein in conjunction with MPTP may create Lewy bodies [115,116]. Hence, systemically administered MPTP in African green monkeys has become acknowledged as the most authentic model for actual human idiopathic PD and the model in which the safety and efficacy of viral vector- and cell-based therapies in humans can most reliably be judged [1,8–10,117]. Indeed, the potential use of lentiviral and adeno-associated virus-based vectors for delivering neurotrophic factors in PD patients has been, and is currently being, tested in monkies [118–120]. Such studies, in turn, set the stage for some Phase I clinical trials [121–131]. It is disheartening that, to date, very few investigators in the cell therapy field (including the stem cell field) have taken advantage of this model for work in PD, despite widespread recognition of its power. This reticence is, most probably, due to the huge expense entailed in performing such complex studies with a sufficiently large sample size and with suitably objective and predictive readouts over a long enough period of observation. Nevertheless, that expense is vastly smaller than the cost to the healthcare system, science and patient well-being when an ineffective or injurious clinical intervention becomes misguidedly launched. Funding agencies and scientists must begin to change their thinking and view adequately powered large animal studies for some diseases not as a luxury, but as a necessity.Another source of hesitancy in the use of large animals, particularly monkeys, is not solely financial, but also ethical. There is no question that experimentation on ‘higher order’ animals provokes, more than any other type of scientific study, circumspection regarding the appropriateness of animal experimentation. Although we have outlined its scientific justification, there is no question that investigators must be acutely sensitive (as they should be for all animal work) in using the minimal number of monkeys required to statistically overcome type 1 and 2 error rates [1], to treating the animals humanely and to only using animals in the most judicious manner and only for exquisitely well-conceived and rigorously designed experiments. Another level of ethical consideration that has recently emerged as a consequence of the burgeoning human stem cell field, is whether integrating human cells into the brains of nonhuman primates (particularly prenatally) somehow ‘humanizes’ them. In other words, might a human–primate chimeric brain develop consciousness and, hence, merit consideration beyond that of just an experimental laboratory animal [131–134]. While an interesting metaphysical consideration, chimerism in our hands, even under the most optimal transplantation conditions, yields only a relatively small human cell contribution to an overwhelmingly primate CNS structure. Furthermore, we have never observed anything but typical primate behaviors, emotions and skills in our animals.Summary & conclusions Appropriately assessing the safety and efficacy of many future interventions in regenerative medicine will require large animal preclinical testing to avoid the failures of previous studies in patients. Such recognition requires scientists, investors, funding agencies and the public to recalibrate their thinking. Simpler rodent models, while useful for unraveling certain biological conundrums, often fall short in their predictive value for human disease or their appropriateness for devising effective delivery methods. A greater number of multicenter collaborations may be required to insure adequate funding and expertise for large animal studies. Such a method requires a less egocentric approach to scientific accomplishment. The ultimate cost of not including the rationale use of large animal models in preclinical studies – the price of unsuccessful trials, injury to patients and failure to bring potentially groundbreaking therapies to the bedside – far outweighs the short-term expense of the studies themselves.AcknowledgementsWe thank DE Redmond and JH Kordower for helpful suggestions. DR Wakeman is supported by the National Institute of Health (NIH)/National Institute of General Medical Sciences (NIGMS) funded UCSD Genetics Training Program (T32 GM08666) and is a recipient of an American Society for Neural Therapy and Repair Travel Fellowship (R13NSO55615; DR Wakeman and AM Crain are supported by an NIH Stem Cell Center Grant (1 P20 GM075059–01) and a grant from the American Parkinson’s Disease Association.DR Wakeman, AM Crain and EY Snyder acknowledge no conflicts of interest.Bibliography1 Redmond DE Jr: Cellular replacement therapy for Parkinson's disease – where we are today? Neuroscientist8(5), 457–488 (2002).Crossref, Medline, CAS, Google Scholar2 Lindvall O, Rehncrona S, Brundin P et al.: Human fetal dopamine neurons grafted into the striatum in two patients with severe Parkinson’s disease: a detailed account of methodology and a 6-month follow-up. Arch. Neurol.46, 615–631 (1989).Crossref, Medline, CAS, Google Scholar3 Bacopoulos NG, Maas JW, Hattox SE, Roth RH: Regional distribution of dopamine metabolites in human and primate brain. Comm. Psychopharmacol.2, 281–286 (1978).Medline, CAS, Google Scholar4 Felten DL, Sladek JRJ: Monoamine distribution in primate brain: V. monoamine nuclei: anatomy, pathways and local organization. Brain Res. Bull.10, 171–284 (1983).Crossref, Medline, CAS, Google Scholar5 Waddington JL, O’Boyle KM: Drugs acting on brain dopamine receptors: a conceptual reevaluation five years after the first selective D1 antagonist. Pharmacol. Ther.43, 1–52 (1989).Crossref, Medline, CAS, Google Scholar6 Haycock JW: Four forms of tyrosine hydroxylase are present in human adrenal medulla. J. Neurochem.56, 2139–2142 (1991). Crossref, Medline, CAS, Google Scholar7 Nagatsu T: Genes for human catecholamine-synthesizing enzymes. Neurosci. Res.12, 315–345 (1991).Crossref, Medline, CAS, Google Scholar8 Steece-Collier K, Maries E, Kordower JH: Etiology of Parkinson's disease: genetics and environment revisited. Proc. Natl Acad. Sci. USA 99(22), 13972–1394 (2002).Crossref, Medline, CAS, Google Scholar9 Collier TJ, Steece-Collier K, Kordower JH: Primate models of Parkinson's disease. Exp. Neurol.183(2), 258–262 (2003).Crossref, Medline, Google Scholar10 Emborg ME: Evaluation of animal models of Parkinson’s disease for neuroprotective strategies. J. Neurosci. Methods 139(2), 121–143 (2004).Crossref, Medline, CAS, Google Scholar11 Taylor JR, Roth RH, Sladek JR Jr, Redmond DE Jr: Cognitive and motor deficits in the performance of an object retrieval task with a barrier-detour in monkeys (Cercopithecus aethiops sabaeus) treated with MPTP: long-term performance and effect of transparency of the barrier. Behav. Neurosci.104(4), 564–576 (1990). Crossref, Medline, CAS, Google Scholar12 Taylor JR, Elsworth JD, Roth RH, Sladek JR Jr, Redmond DE Jr: Cognitive and motor deficits in the acquisition of an object retrieval/detour task in MPTP-treated monkeys. Brain113(Pt 3), 617–637 (1990).Crossref, Medline, Google Scholar13 Taylor JR, Elsworth JD, Roth RH, Collier TJ, Sladek JR, Redmond DE Jr: Improvements in MPTP-induced object retrieval deficits and behavioral deficits after fetal nigral grafting in monkeys. In: Progress in brain research. Richards SJ, Dunnett SB (Eds). Elsevier, Amsterdam, The Netherlands 543–559 (1990).Crossref, Google Scholar14 Elsworth JD, Lawrence MS, Roth RH et al.: D1 and D2 dopamine receptors independently regulate spontaneous blink rate in the vervet monkey. J. Pharmacol. Exp. Ther.259, 595–600 (1991).Medline, CAS, Google Scholar15 Taylor JR, Elsworth JD, Roth RH, Sladek JR Jr, Collier TJ, Redmond DE Jr: Grafting of fetal substantia nigra to striatum reverses behavioral deficits induced by MPTP in primates: a comparison with other types of grafts as controls. Exp. Brain Res.85(2), 335–348 (1991).Crossref, Medline, CAS, Google Scholar16 Kordower JH, Liu YT, Winn S, Emerich DF: Encapsulated PC12 cell transplants into hemiparkinsonian monkeys: a behavioral, neuroanatomical, and neurochemical analysis. Cell Transplant.4(2), 155–171 (1995).Crossref, Medline, CAS, Google Scholar17 Vodicka P, Smetana K Jr, Dvorankova B et al.: The miniature pig as an animal model in biomedical research. Ann. N. Y. Acad. Sci.1049, 161–171 (2005).Crossref, Medline, Google Scholar18 Bollen P, Ellegaard L: Developments in breeding Göttingen minipigs. In: Advances in Swine in Biomedical Research. Tumbleson ME, Schook LB (Eds). Plenum Press, NY, USA 59–66 (1996).Google Scholar19 Bollen P, Ellegaard L: The Göttingen minipig in pharmacology and toxicology. Pharmacol. Toxicol.80, 3–4 (1997).Crossref, Medline, CAS, Google Scholar20 Brown D, Terris J: Swine in physiological and pathophysiological research. In: Advances in Swine in Biomedical Research. Tumbleson ME, Schook LB (Eds). Plenum Press, NY, USA 5–6 (1996).Google Scholar21 Murakami T, Hitomi S, Ohtsuka A, Taguchi T, Fujita T: Pancreatic insulo-acinar portal systems in humans, rats, and some other mammals: Scanning electron microscopy of vascular casts. Microsc. Res. Tech.37, 478–488 (1997).Crossref, Medline, CAS, Google Scholar22 Larsen MO, Rolin B: Use of the Göttingen minipig as a model of diabetes, with special focus on type 1 diabetes research. ILAR J. 45(3), 303–313 (2004).Crossref, Medline, CAS, Google Scholar23 Markussen J, Havelund S, Kurtzhals P et al.: Soluble, fatty acid acylated insulins bind to albumin and show protracted action in pigs. Diabetologia39, 281–288 (1996).Crossref, Medline, CAS, Google Scholar24 Pieber TR, Plank J, Görzer E et al.: Duration of action, pharmacodynamic profile and between-subject variability of insulin determine subjects with type 1 diabetes. Diabetologia45, A257 (2002).Google Scholar25 Miller ER, Ullrey DE: The pig as a model for human nutrition. Annu. Rev. Nutr. 7, 361–382 (1987).Crossref, Medline, CAS, Google Scholar26 Broughton DL, Taylor R: Review: deterioration of glucose tolerance with age: the role of insulin resistance. Age Ageing20, 221–225 (1991).Crossref, Medline, CAS, Google Scholar27 Rosenthal M, Doberne L, Greenfield M, Widstrom A, Reaven GM: Effect of age on glucose tolerance, insulin secretion, and in vivo insulin action. J. Am. Geriatr. Soc.30, 562–567 (1982).Crossref, Medline, CAS, Google Scholar28 Marshall M, Sprandel U, Zollner N: Streptozotocin diabetes in a miniature pig. Res. Exp. Med.165, 61–65 (1975).Crossref, Medline, CAS, Google Scholar29 Marshall M: Induction of chronic diabetes by streptozotocin in the miniature pig. Res. Exp. Med.175, 187–196 (1979).Crossref, Medline, CAS, Google Scholar30 Gabel H, Bitter-Suermann H, Henriksson C, Save-Soderbergh J, Lundholm K, Brynger H:Streptozotocin diabetes in juvenile pigs. Evaluation of an experimental model. Horm. Metab. Res.17, 275–280 (1985).Crossref, Medline, CAS, Google Scholar31 Wilson JD, Dhall DP, Simeonovic CJ, Lafferty KJ: Induction and management of diabetes mellitus in the pig. Aust. J. Exp. Biol. Med. Sci.64, 489–500 (1986). Crossref, Medline, Google Scholar32 Kjems LL, Kirby BM, Welsh EM et al.: Decrease in ??cell mass leads to impaired pulsatile insulin secretion, reduced postprandial hepatic insulin clearance, and relative hyperglucagonemia in the minipig. Diabetes50, 2001–2012 (2001).Crossref, Medline, CAS, Google Scholar33 Mellert J, Hopt UT, Hering BJ, Bretzel RG, Federlin K: Influence of islet mass and purity on reversibility of diabetes in pancreatectomized pigs. Transplant Proc.23, 1687–1689 (1991).Medline, CAS, Google Scholar34 Morsiani E, Fogli L, Lanza G, Lebow LT, Demetriou AA, Rozga J: Long-term insulin independence following repeated islet transplantation in totally pancreatectomized diabetic pigs. Cell Transplant.11, 55–66 (2001).Crossref, Google Scholar35 Kin T, Iwata H, Aomatsu Y et al.: Xenotransplantation of pig islets in diabetic dogs with use of a microcapsule composed of agarose and polystyrene sulfonic acid mixed gel. Pancreas25, 94–100 (2002).Crossref, Medline, Google Scholar36 Buhler L, Deng S, O’Neil J et al.: Adult porcine islet transplantation in baboons treated with conventional immunosuppression or a nonmyeloablative regimen and CD154 blockade. Xenotransplantation9, 3–13 (2002).Crossref, Medline, CAS, Google Scholar37 Cantarovich D, Blancho G, Potiron N et al.: Rapid failure of pig islet transplantation in non human primates. Xenotransplantation9, 25–35 (2002).Crossref, Medline, Google Scholar38 Groth CG, Korsgren O, Tibell A et al.: Transplantation of porcine fetal pancreas to diabetic patients. Lancet344, 1402–1404 (1994).Crossref, Medline, CAS, Google Scholar39 Elliott RB, Escobar L, Garkavenko O et al. No evidence of infection with porcine endogenous retrovirus in recipients of encapsulated porcine islet xenografts. Cell Transplant.9, 895–901 (2000).Crossref, Medline, CAS, Google Scholar40 Groth CG, Tibell A, Wennberg L et al.: Clinical aspects and perspectives in islet xenotransplantation. J. Hepatobil. Pancreat. Surg.7, 364–369 (2000).Crossref, Medline, CAS, Google Scholar41 Shapiro AMJ, Lakey JRT, Ryan EA, et al.: Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N. Engl. J. Med.343, 230–238 (2000).Crossref, Medline, CAS, Google Scholar42 Sachs D, Leight HG, Cone J, Schwarz S, Stuart L, Rosenberg S: Transplantation in miniature swine. I. Fixation of the major histocompatability complex. Transplantation22, 559–567 (1976).Crossref, Medline, CAS, Google Scholar43 Sachs DH: MHC-homozygous miniature swine. In: Swine as Models in Biomedical Research. Swindel MM, Moody DC, Philips LD (Eds). Iowa State University Press, IA, USA 3–15 (1992).Google Scholar44 Mezrich JD, Haller GW, Arn JS, Houser SL, Madsen JC, Sachs DH. Histocompatible miniature swine: an inbred large-animal model. Transplantation, 75(6), 904–907 (2003).Crossref, Medline, Google Scholar45 Allan JS, Wain JC, Schwarze ML et al.: Modeling chronic lung allograft rejection in miniature swine. Transplantation 73(3), 447–453 (2002).Crossref, Medline, Google Scholar46 Gollackner B, Dor FJ, Knosalla C et al.: Spleen transplantation in miniature swine: surgical technique and results in major histocompatibility complex-matched donor and recipient pairs. Transplantation 75(11), 1799–806 (2003).Crossref, Medline, CAS, Google Scholar47 Kenmochi T, Mullen Y, Miyamoto M, Stein E: Swine as an allotransplantation model. Vet. Immunol. Immunopathol.43(1–3), 177–183 (1994).Crossref, Medline, CAS, Google Scholar48 Emery DW, Sachs DH, LeGuern C: Culture and characterization of hematopoietic progenitor cells from miniature swine. Exp. Hematol.24(8), 927–935 (1996).Medline, CAS, Google Scholar49 Tumbleson ME: Brain weight, as a function of age, in miniature swine. Growth37(1), 13–17 (1973).Medline, CAS, Google Scholar50 Holloway RL, Heilbroner P: Corpus callosum in sexually dimorphic and nondimorphic primates. Am. J. Phys. Anthropol.87(3), 349–357 (1992). Crossref, Medline, CAS, Google Scholar51 Singer BA, Tresser NJ, Frank JA, McFarland HF, Biddison WE: Induction of experimental allergic encephalomyelitis in the NIH minipig. J. Ieuroimmunol. 105(1), 7–19 (2000).Crossref, Medline, CAS, Google Scholar52 Matsuyama N, Hadano S, Onoe K et al.: Identification and characterization of the miniature

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call